You are on page 1of 13

&get_box_var;

ORIGINAL ARTICLE

Microbiota Promotes Chronic Pulmonary Inflammation by Enhancing


IL-17A and Autoantibodies
Koshika Yadava1,2, Céline Pattaroni1, Anke K. Sichelstiel1, Aurélien Trompette1, Eva S. Gollwitzer1, Olawale Salami1,
Christophe von Garnier3,4, Laurent P. Nicod1, and Benjamin J. Marsland1
1
Service de Pneumologie, Faculté de Biologie et de Médecine, Centre Hospitalier Universitaire Vaudoise–Université Lausanne
Lausanne, Switzerland; 2Division of Infectious Diseases, Department of Medicine, Stanford University School of Medicine, Stanford,
California; 3Department of Respiratory Medicine, Inselspital, Bern University Hospital, Bern, Switzerland; and 4Department of Clinical
Research, University of Bern, Bern, Switzerland

Abstract richness and diversity were decreased in LPS/elastase–treated


mice, with an increased representation of the genera
Rationale: Changes in the pulmonary microbiota are associated Pseudomonas and Lactobacillus and a reduction in Prevotella.
with progressive respiratory diseases including chronic obstructive Moreover, the microbiota was implicated in disease development
pulmonary disease (COPD). Whether there is a causal relationship as mice depleted, or devoid, of microbiota exhibited an
between these changes and disease progression remains unknown. improvement in lung function, reduced inflammation, and
lymphoid neogenesis. The absence of microbial cues markedly
Objectives: To investigate the link between an altered microbiota decreased the production of IL-17A, whereas intranasal
and disease, we used a murine model of chronic lung inflammation transfer of fluid enriched with the pulmonary microbiota
that is characterized by key pathological features found in isolated from diseased mice enhanced IL-17A production
COPD and compared responses in specific pathogen–free (SPF) in the lungs of antibiotic-treated or axenic recipients.
mice and mice depleted of microbiota by antibiotic treatment or Finally, in mice harboring a microbiota, neutralizing
devoid of a microbiota (axenic). IL-17A dampened inflammation and restored lung
Methods: Mice were challenged with LPS/elastase intranasally over function.
4 weeks, resulting in a chronically inflamed and damaged lung.
The ensuing cellular infiltration, histological damage, and decline Conclusions: Collectively, our data indicate that host–microbial
in lung function were quantified. cross-talk promotes inflammation and could underlie the chronicity
of inflammatory lung diseases.
Measurements and Main Results: Similar to human
disease, the composition of the pulmonary microbiota was
altered in diseased animals. We found that the microbiota Keywords: microbiome; COPD; autoimmunity; IL-17

The rise in the prevalence of chronic presents as chronic bronchitis and increased severity and progression remain
obstructive pulmonary disease (COPD) emphysema that lead to progressive and unclear (3, 5). Despite the heterogeneity in
is a global health concern (1). Exposure irreversible airflow limitation (3, 4). human disease, it is apparent that aberrant
to cigarette smoke is the most widely Although patients can manifest mild to inflammatory responses substantially
associated environmental risk factor for the severe disease, as defined by the degree of contribute to the decline in lung function
development of the disease (2), which airflow obstruction, the signals leading to (5–7).

( Received in original form April 19, 2015; accepted in final form December 1, 2015 )
Funded by the Swiss National Science Foundation grant CRSII3_141875 (B.J.M. and C.v.G). K.Y. is currently supported by the Swiss National Science
Foundation early postdoc mobility grant.
Author Contributions: Conceived and designed the experiments: K.Y. and B.J.M. Performed the experiments and analyzed data: K.Y., A.K.S., A.T., O.S.,
E.S.G., C.P., and B.J.M. Prepared the manuscript: K.Y., B.J.M., L.P.N., and C.v.G.
Correspondence and requests for reprints should be addressed to Koshika Yadava, Ph.D., Division of Infectious Diseases, Department of Medicine, Beckman
Building B237, 279 Campus Drive, Stanford University School of Medicine, Stanford, CA 94305. E-mail: koshika@stanford.edu
This article has an online supplement, which is accessible from this issue’s table of contents at www.atsjournals.org
Am J Respir Crit Care Med Vol 193, Iss 9, pp 975–987, May 1, 2016
Copyright © 2016 by the American Thoracic Society
Originally Published in Press as DOI: 10.1164/rccm.201504-0779OC on December 2, 2015
Internet address: www.atsjournals.org

Yadava, Pattaroni, Sichelstiel, et al.: Microbiota Sustains Chronic Pulmonary Inflammation 975
ORIGINAL ARTICLE

have been consistently observed in both tracheotomized and mechanically


At a Glance Commentary animal models and humans with COPD ventilated at a rate of 450 breaths/min and a
(24–27), there are conflicting reports tidal volume of 10 ml/kg bodyweight.
Scientific Knowledge on the regarding the production of self-reactive
Subject: The airway microbiota is antibodies (28–32); thus, the autoimmune Antibiotic Treatment
different in healthy individuals and concept of COPD remains open and might For depleting microbiota before the start
patients with chronic obstructive reflect a subtype of the disease. Incidentally, of the experiment, mice were given 10%
pulmonary disease. However, its cues from the microbiota have been enrofloxacin (Baytril) in drinking water for
function is unknown. reported to support lymphoid neogenesis 2 weeks followed by amoxicillin/clavulanic
(33) and the production of self-reactive acid (Co-Amoxi-Mepha) in the drinking
What This Study Adds to the antibodies (34) and are implicated in the water for another 2 weeks. During the course
Field: This study provides pathogenesis of a variety of autoimmune of the experiment mice were maintained on
mechanistic insight into the function of models (34–36). Whether there could be a Co-Amoxi-Mepha treatment.
the airway microbiota in a mouse similar link between the pulmonary 16S rDNA preparation from
model of chronic obstructive microbiota and autoimmune mechanisms bronchoalveolar lavage fluid and
pulmonary disease. in COPD is yet to be established. sequencing. Bronchoalveolar lavage (BAL)
We aimed to elucidate the role of the was performed in 1 ml volume of sterile
microbiome in COPD using a murine model phosphate-buffered saline (PBS) and
Intriguingly, not all people exposed to (37), which mimics key features of human collected in a 2 ml Biopure tube (Eppendorf,
similar levels of cigarette smoke develop disease. We found that akin to what has Basel, Switzerland). Negative control was
the disease (3). Moreover, although been seen in humans, the airway microbiota obtained by flushing 1 ml of sterile PBS into
smoking cessation does lead to an differs between healthy and diseased mouse the BAL tubing system and processed
improvement in lung function in moderate lungs. We report that the microbiota has identically to the mouse BAL fluid (BALF)
disease, in some cases it does not impact functional implications, as in its absence samples. BALF was directly centrifuged at
disease progression (8). One could disease development is abrogated. 14,000 3 g for 4 minutes at 48 C and
speculate that although environmental processed in sterile conditions under a
triggers enhance the disease, in susceptible laminar flow hood. Pellets were incubated
individuals the established disease can Methods with 9,000 U of Ready-Lyse Lysozyme
progress through a self-sustained (Epicentre, Hessisch Oldendorf, Germany)
inflammatory cycle. Given the limitations Mice for 1 hour at 378 C. Twenty microliters of
of current therapies to arrest COPD BALB/c mice were purchased from Charles proteinase K in 200 ml AL buffer (QIAamp
progression, it is important to identify the River, l’Arbresele, France. All animal DNA Mini Kit) was then added to the
factors that can facilitate the establishment experiments were performed according to solution and incubated for 30 minutes at
of the chronic inflammatory pathways and institutional guidelines and Swiss federal 568 C with shaking. After this step, BALF
favor the disease. One such factor could be and cantonal laws on animal protection. from four mice coming from two different
the microbiota. Previously considered sterile, cages were pooled and further processed with
breakthroughs in non–culture-based Model of Chronic Pulmonary QIAmp DNA Mini Kit, according to the
detection methods have provided evidence Inflammation manufacturer’s protocol. DNA was eluted
that the respiratory tract harbors a Mice were exposed intranasally to a mixture in 30 ml DNase/RNAse-free water (Sigma-
microbiota (9–18). Notably, the microbiota of 7 mg LPS from Escherichia coli O26:B6 Aldrich). Polymerase chain reaction (PCR)
in the respiratory tract of healthy volunteers, (Sigma-Aldrich, St. Gallen, Switzerland) for the 16S rDNA library preparation was
smokers without COPD, and patients with and 1.2 U porcine pancreatic elastase performed using modified 27F and 338R
severe disease are distinct (9, 10, 12–14). (Elastin Products Company, Owensville, universal primers, which target the V1-V2
Although microbial dysbiosis has been MO) in 100 ml once a week over 4 weeks. hypervariable region of the 16S rDNA gene.
shown to increase susceptibility to chronic Terminal readout was performed 1 week Primers were as follows:
diseases such as inflammatory bowel disease after the last challenge.
27F:
(19, 20), it is currently unknown whether
59-AATGATACGGCGACCACC
such a phenomenon contributes to COPD. Assessment of Pulmonary Lung
GAGATCTACACTATGGTAATTCC
Another emerging hypothesis suggests Function
AGMGTTYGATYMTGGCTCAG-39
there is an autoimmune component driving Lung compliance and FEV/FVC parameters
and
the progression of COPD (5, 21, 22). were measured by Snapshot and NPFE
338R:
Although controversial, there is both perturbations using the FlexiVent invasive
59-CAAGCAGAAGACGGCA
circumstantial and direct evidence airway mechanics system from Scireq
TACGAGATNNNNNNNNNNNN
implicating autoimmune mechanisms in (Montreal, Canada). Mice were anesthetized
AGTCAGTCAGAAGCTGCCTCCCG
COPD pathogenesis (5, 21, 22). Lymphoid by administering 100 mg/kg ketamine
TAGGAGT-39,
follicles play a pathological role in several (Ketasol-100; Graeub) intramuscularly and
autoimmune diseases by mediating the 50 mg/kg pentobarbital (Esconarkon; where Illumina adaptor sequences
generation of self-reactive antibodies (23). Streuli Pharma, Uznach, Switzerland) are bold, linkers are italicized, and the
Although lymphoid follicles themselves intraperitoneally. Subsequently, mice were NNNNNNNNNNNN sequence represents

976 American Journal of Respiratory and Critical Care Medicine Volume 193 Number 9 | May 1 2016
ORIGINAL ARTICLE

the sample-specific molecular identifier tag Preparation and Transfer of (Figure 1I). Notably, the emphysema and
barcodes. The temperature cycles were set as Microbiota-enriched BAL Fluid inflammation persisted at least 2 months
follows: 3 minutes of initial denaturation at For preparing the microbiota-enriched BAL after the cessation of LPS/elastase
948 C, 40 cycles of 30 seconds of denaturation fluid, BAL was performed three times using treatment, (P , 0.001) (see Figures
at 948 C, 30 seconds of annealing at 568 C, 1 ml of PBS per mouse. Five mice were E1A–E1C in the online supplement).
1.3 minutes of extension at 728 C, and a final pooled per group. The BAL was then Although neutrophilia waned in the BAL,
step of 5 minutes at 728 C. PCRs were centrifuged at 2,000 rpm for 10 minutes at the continued prevalence of hemosiderin-
performed in triplicate, and one additional 48 C, and the pellet containing mouse laden macrophages (P , 0.01) was
PCR negative control without DNA cells was discarded. The BAL supernatant indicative of chronic inflammation and
template was added for each reaction. Each was aliquoted into Biopur tubes and microhemorrhaging months after the final
reaction consisted of 8 ml of DNA template, centrifuged at 14,000 3 g for 10 minutes challenge (Figure E1D) (42).
0.44 ml of each 27F and 338R primers at at 48 C and washed with PBS at the same
10 mM, 2 ml of AccuPrime buffer II, 0.09 ml speed. The final pellet was snap frozen in Chronic Pulmonary Inflammation Is
of AccuPrime Taq DNA polymerase high liquid nitrogen and stored at 2808 C until Associated with Lymphoid
fidelity (Invitrogen), and DNase/RNAse- use. Before intranasal administration, Neogenesis, Enhanced Local
free water (Sigma-Aldrich) to reach a total each aliquot was resuspended in 2 ml of Antibody Responses, and IL-17A
volume of 20 ml. After amplification, PCR PBS and administered to recipient mice in Production
product triplicates were pooled before a volume of 100 ml per mouse In line with previous data in human
visualization on a 1.5% agarose gel. The intranasally. disease and animal models of cigarette
bands of the specific size (approximately smoke–induced emphysema (24–27), we
300 bp) were cut and purified with QIAquick Neutralization of IL-17A observed the formation of lymphoid
gel extraction kit (Qiagen, Hombrechtikon, For neutralization of IL-17A, mice were follicles (LFs), which could be categorized
Switzerland). Recovered DNA was then treated with 250 mg of anti–IL-17A as bronchiolar-associated lymphoid tissue
quantified using the Quant-iT PicoGreen ds (clone 17F3) or the corresponding isotype (P , 0.01), alveolar-associated lymphoid
assay (Life Technologies, ThermoFisher, control antibody (clone MPOC-21) from tissue (P , 0.01), or vascular-associated
Waltham, MA), and amplicons were pooled BioXCell (West Lebanon, NH). Antibodies lymphoid tissue (VALT) (P , 0.001) based
in a single tube in equimolar amounts. were administered intraperitoneally on on their localization (Figure 2A). We found
Sequencing was performed on an Illumina Days 6, 10, 13, 17, 20, and 24 after the first that VALT accounted for the majority of
MiSeq platform using MiSeq reagent kit challenge of LPS/elastase challenge. LFs. Moreover, these LFs persisted even in
V2-500 (pair-end, 2 3 250). the absence of inflammatory stimuli in mice
16S rDNA sequencing analysis. that were analyzed 9 weeks after the last
Sequences obtained were processed and Results challenge (P , 0.05) (Figure E1E). Overall,
analyzed using Quantitative Insights into there was an increase in the frequency
Microbial Ecology (QIIME, v.1.8.0) software Murine Model of Chronic Pulmonary of germinal center B cells (P , 0.05)
(38). Paired forward and reverse reads were Inflammation Mimics Key (Figure 2B) as identified by Gl7 staining
merged using fastq-join, demultiplexed, Pathological Features of Human and T follicular helper cells (P , 0.01)
and quality filtered (quality Phred score Disease (Figure 2C) as defined by the coexpression
Q . 20, fewer than three low-quality base To study the underlying pathogenic of PD-1 (programmed cell death protein 1)
calls). Operational taxonomic units (OTUs) mechanisms of COPD, we used a murine and Gl7 in the lungs of LPS/elastase–treated
were assigned using an open reference model of chronic pulmonary inflammation mice. To assess whether the presence of LFs
picking strategy with Uclust (39) at 97% (Figure 1A). Mice were treated with a correlated with an increase in the local
identity against the 97% Greengenes combination of LPS and elastase over antibody response, we measured the
reference database (v13.8) (40). Alpha 4 weeks. Coadministration of LPS/elastase amount of the antibody isotypes IgG1,
diversity rarefaction curves for chao1 and resulted in an increase in compliance, IgA, and IgM in the BAL of PBS- or
Shannon indexes were calculated in QIIME (P , 0.05) (Figure 1B) and a decrease in LPS/elastase–treated mice. All isotypes were
using 10 iterations represented as mean 1 the percentage of FEV0.1/FVC (P , 0.01) increased in the BAL of LPS/elastase–
SD at different sequencing depths (Figure 1C). The decline in lung function treated mice (P , 0.05) (Figure 2D). Of
(2,000–98,000). All downstream analyses was associated with emphysematous note, no increase in the systemic levels of
were performed using a rarefied OTU table changes in the lung parenchyma, quantified any isotype was detected (Figure E2A).
at 40,000 sequencing depth. A heatmap by both mean linear intercept and We then investigated the reactivity of
displaying the OTUs represented at more destruction index parameters (P , 0.001) these antibodies against collagen and elastin,
than 0.05% relative abundance in a (Figures 1D–1F) in addition to which constitute a component of the lung
minimum of two samples was created using peribronchiolar and perivascular tissue breakdown products. A significant
the heatmap.2 function of the gplots infiltration of the lung tissue (Figure 1G). increase in specific responses against
package in R (41). The dendrogram was There was also an enhanced cellular collagen was detected for IgG1, (P , 0.01)
generated using Ward’s hierarchical infiltration in the BALF (P , 0.001) and IgA (P , 0.001) but not IgM isotypes
clustering with hclust algorithm in R on (Figure 1H), with marked increase in (Figures 2E–2G). A marked increase in
Bray-Curtis dissimilarity matrix calculated macrophages (P , 0.05), neutrophils specific antibodies against elastin was
in QIIME. (P , 0.05), and lymphocytes (P , 0.001) observed for IgA (P , 0.05), IgG1

Yadava, Pattaroni, Sichelstiel, et al.: Microbiota Sustains Chronic Pulmonary Inflammation 977
ORIGINAL ARTICLE

A B *
C **
PBS or

Compliance (cm H2O.s/ml)


LPS/elastase (L/E) 0.10 100
intranasally

FEV 0.1/FVC (%)


0.08
80

0.06
Week 1 2 3 4 5
60

Final Analysis PBS LPS/elastase PBS LPS/elastase

D E F
*** ****

Mean linear intercept(m)


PBS LPS/elastase 150 100

Destruction index
80
100
60

40
50
20

PBS LPS/elastase PBS LPS/elastase

G H I
PBS LPS/elastase 8
*** 6.5 PBS

Number in BAL(X105)
Total cells in BAL(X105)

* * LPS/elastase
4.5
6
**
2.5
4
0.5
2 0.2
0.1

PBS LPS/elastase Mac Neu Lym Eos

Figure 1. Murine model of chronic pulmonary inflammation mimics key pathological features of human disease. (A) BALB/c mice were administered
LPS (7 mg) and elastase (1.5 U) in a volume of 100 ml intranasally once a week for 4 weeks. Terminal analysis was performed 1 week after the last
challenge. (B) Lung compliance and (C) FEV0.1/FVC were measured by FlexiVent invasive airway mechanics system. (D) Paraformaldehyde-fixed
sections of lung were stained with hematoxylin and eosin, and emphysema and destruction of lungs were scored by (E) mean linear intercept and (F)
destruction index (n = 3–5). (G) Representative hematoxylin and eosin–stained lung sections showing perivascular and peribronchiolar inflammation
in LPS/elastase–treated mice. (H) The total cell and (I) differential counts in the bronchoalveolar alveolar lavage (BAL) were determined (n = 5). Error bars
represent SEM. Data representative of at least two independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. Eos = eosinophils;
Lym = lymphocytes; Mac = macrophages; Neu = neutrophils; PBS = phosphate-buffered saline.

(P , 0.01), and IgM (P , 0.05) antibody collagen or elastin in vitro. Furthermore, we LPS/elastase over 4 weeks and assessed
isotypes (Figures 2E–2G). We did not found that the protein levels of IL-6 and microbiota diversity and composition by
detect an increase in systemic levels of IL-1b, cytokines that can enhance IL-17A Illumina 16S rDNA sequencing using a
elastin-specific IgG1 and IgA responses, responses, in addition to levels of IL-17A custom protocol. Indeed, amplifying
although collagen-specific antibodies were itself, were increased in the BALF of bacterial DNA from mouse BAL is
increased (Figures E2B and E2C). To LPS/elastase–treated animals (Figures particularly challenging given the low
further characterize the autoimmune E3A–E3C) biomass and subsequent high risk of
aspect of the disease, we enumerated contamination. For these reasons, we
IL-17–producing T cells (Figures 2H–2K). We The Pulmonary Microbiota Is Altered pooled BAL from four mice to increase the
found that IL-17 production by both gd1 upon the Induction of Chronic bacterial biomass, and we used stringent
(P , 0.0001) (Figure 2H) and CD41 T cells Pulmonary Inflammation negative controls where PBS (as used for
(P , 0.0001) was increased in the diseased It has been reported that humans with the BAL) was exposed to the same
lungs (Figure 2I). IL-17A production was COPD have an altered airway microbiota environment, tubing, and extraction kits as
also induced when sorted gd1 (Figure 2J) (10, 13); thus, we investigated whether the BAL from mice. A negative control,
and CD41 T cell (Figure 2K) populations similar changes occurred in our mouse where we successfully found an amplified
isolated from the lungs of LPS/elastase- model. We extracted bacterial DNA bacterial 16S rDNA fragment, was included
treated mice were restimulated with from BAL of mice treated with PBS or in the sequencing analyses to allow

978 American Journal of Respiratory and Critical Care Medicine Volume 193 Number 9 | May 1 2016
ORIGINAL ARTICLE

A BALT VALT ALT 20 ****

No./section of the lung


PBS
LPS/elastase
15
**
10
**
5
100 μm 100 μm 100 μm
0
VALT BALT ALT

B GC B cells C Tfh cells

No.in the lung (X104)


No.in the lung (X105)

PBS LPS/elastase 4 * PBS LPS/elastase 4 ***


3 3
2 2
GL-7

GL-7
1 1
0 0
PD-1 PBS LPS/elastase PD-1 PBS LPS/elastase

D Total antibodies in BAL E Specific IgG1 F Specific IgA G Specific IgM


50 0.08 ** ** 0.05 *** 0.6 *
** PBS *
40 LPS/elastase 0.06 0.04
0.4
30 0.03
μg/ml

μg/ml
μg/ml

μg/ml
0.04
20 0.02
**** 0.2
0.02
10 *** 0.01
nd nd
0
IgG1 IgA IgM Collagen Elastin Collagen Elastin Collagen Elastin

H γδ+ T cells I
**** Media Collagen Elastin
PBS LPS/elastase 80
0.74% 2.48% 3.46%
%IL-17A+ of
γδ+ T cells

60
40
IL-17A
IL-17A

20

IFNγ PBS LPS/elastase γδ

J CD4+ T cells **** K Media Collagen Elastin


40
PBS LPS/elastase 0.091% 0.44% 0.36%
CD4+ T cells
%IL-17A+ of

30
20
IL-17A

IL-17A

10

IFNγ PBS LPS/elastase CD4


Figure 2. Chronic pulmonary inflammation is associated with lymphoid neogenesis, enhanced local antibody responses, and IL-17A production. (A)
Representative pictures of bronchial-associated lymphoid tissue (BALT), vessel-associated lymphoid tissue (VALT), and alveolar-associated lymphoid
tissue (ALT) in hematoxylin and eosin–stained lung sections and their quantification. Data pooled from two experiments (n = 6–8). (B) Germinal center (GC)
B cells and (C) follicular T helper (Tfh) cells from lungs were quantified by flow cytometry. Data pooled from two experiments. (D) Levels of IgG1, IgA,
and IgM in bronchoalveolar lavage (BAL) supernatant were measured by ELISA. Data pooled from two experiments (n = 8–9). Collagen- and elastin-
specific (E) IgG1, (F) IgA, and (G) IgM responses were quantified by ELISA. Data pooled from two experiments (n = 8–9). (H) Cells isolated from lungs and
airways of indicated groups were stimulated in vitro with phorbol myristate acetate, ionomycin, and monensin for 4 hours. IL-17 production by gd1 T cells
was quantified by flow cytometry. Data are representative of at least three independent experiments. (I) IL-17 production by CD41 T cells was quantified by

Yadava, Pattaroni, Sichelstiel, et al.: Microbiota Sustains Chronic Pulmonary Inflammation 979
ORIGINAL ARTICLE

conclusions to be drawn from low- Microbiota-derived Signals Amplify sources of IL-17A, we found no difference
abundance bacterial sequences. We found Innate and Adaptive Inflammation in in the composition of cells producing IL-
that microbiota richness and diversity Disease 17A (Figure 5B) using the gating strategy
were decreased in the pooled BAL of To investigate whether microbiota- shown in Figure E5B, and both CD41 and
LPS/elastase–treated mice, as indicated by dependent signals contribute to the gd1 T cells accounted for the majority
chao1 richness and Shannon diversity persistent inflammation, we compared of IL-17A producers. However, the
rarefaction curves (Figure 3A). Of note, the responses between SPF mice and mice frequencies of IL-17A1 within CD41
Shannon index diversity was higher in the depleted of a microbiota using an antibiotic T cells and the total number of IL-17A1
negative control than the LPS/elastase treatment regime. Before the start of the CD41 T cells were markedly reduced upon
group, potentially indicative of an increased experiment mice were given 10% Baytril in antibiotic treatment (P , 0.01), whereas
abundance of certain bacteria in the drinking water for 2 weeks followed by only a minor reduction was seen for
LPS/elastase group, which is reflected by treatment with Co-Amoxi-Mepha for gd1 T cells, which was not statistically
the abundance component of the Shannon another 2 weeks. This treatment decreased significant (Figures 5C and 5D). Similar
diversity calculation. We next assessed the microbial load by approximately 6 log, to antibiotic-treated mice, axenic mice
microbiota composition, with particular (P , 0.0001) (Figure E4). During the course exhibited reduced recruitment of
attention to the negative control. We of the experiment mice were maintained on neutrophils and macrophages in the
observed that the most abundant Co-Amoxi-Mepha treatment. We found airways (P , 0.05, P , 0.01, respectively)
OTUs present in BAL samples, such that antibiotic-treated mice showed no (Figures E6A–E6D) and a specific reduction
as family Comamonadaceae, family change in FEV0.1/FVC (Figure 4A) but a in IL-17A-producing gd1 T cells (P , 0.05)
Flavobacteriaceae, and family significant reduction in lung compliance (Figure E6E).
Microbacteriaceae were also predominant (P , 0.05) (Figure 4B). This was linked to a In an attempt to assess whether the
in the negative control, but their relative decrease in inflammation in the lungs and altered microbiota in disease could directly
abundance did not differ among BAL airways (Figure 4C), decreased total cell enhance IL-17A responses, we performed a
samples (Figure 3B). Importantly, counts (P , 0.01) (Figure 4D), and transfer experiment. Antibiotic-treated mice
Ward hierarchical clustering algorithm neutrophil (P , 0.01) and interstitial were challenged with LPS/elastase on Days
using Bray-Curtis distance efficiently macrophage (P , 0.01) infiltration in the 0, 7, and 21. Concurrently, they were given a
distinguished and clustered BAL samples of BAL (Figures 4E and 4F). Macrophage solution that was enriched for the airway
the treatments, as compared with the subsets were identified as described in microbiota of PBS- or LPS/elastase–treated
negative control (Figure 3B). These data Figure E5A. Antibiotic treatment also mice intranasally. We found that the
show that a distinct microbiota was present resulted in a reduction in the frequency of transfer of this microbiota-enriched
in healthy airways, diseased airways, and VALT (P , 0.05) (Figure 4G). Although solution from LPS/elastase–treated mice
the background control. To further the total antibody levels of IgG1 and IgM in resulted in an increase in the IL-
investigate the changes in microbiota the BAL were unaffected in antibiotic- 17A–producing cells in the lungs of
induced by LPS/elastase treatment, we treated mice (Figure 4H), collagen-specific recipient mice (Figure 5E). In particular,
excluded contaminant OTUs and assessed IgG1 responses were significantly reduced IL-17A–producing CD41 T cells
the differentially abundant genera across (P , 0.01) (Figure 4I). Comparatively, total (Figure 5F) were increased (P , 0.05),
the treatments. We identified four genera of IgA responses were reduced (P , 0.01) whereas gd1 T cells (Figure 5G) were
particular note: Prevotella, Pseudomonas, (Figure 4H), and, in line with this, collagen- unaffected. Performing the same
Lactobacillus, and Chryseobacterium and elastin-specific IgA responses were experiment using axenic recipients, we
(Figure 3C). Prevotella was present in the reduced (P , 0.05) (Figure 4J). observed an increase in the proportion of
BAL of the PBS control group but absent in IL-17A–producing CD41 and gd1 T cells
the BALF of LPS/elastase–treated mice. Microbiota Enhances Production of (P , 0.05) (Figure E6F).
On the contrary, relative abundance of IL-17A by T Cells
Pseudomonas, Lactobacillus, and Microbiota-dependent signals have been IL-17A Kinetics Correlates with
Chryseobacterium was increased in the shown to amplify autoimmune diseases Disease Severity, and Targeted
BAL of LPS/elastase–treated mice. Overall, in an IL-17–dependent manner (34–36). Neutralization of IL-17A Ameliorates
as reported for the human airway Both autoimmune mechanisms and IL-17 Disease
microbiota, the airways of mice with this (31, 46–54) are implicated in the pathogenesis We next sought to determine the kinetics of
chronic lung disease are characterized by a of COPD. Antibiotic-treated mice showed IL-17A production during the induction of
distinct microbiota to those with healthy a reduced IL-17A response (P , 0.05) disease. We modified our existing model to
airways. (Figure 5A). Upon investigation of the cellular recapitulate different levels of severity

Figure 2. (Continued). flow cytometry in cells isolated from lungs and airways of indicated groups stimulated in vitro with phorbol myristate acetate,
ionomycin, and monensin for 4 hours. Data are representative of at least three independent experiments. (J) Fluorescence-activated cell sorted gd1 T cells
from pooled lungs and airway samples of five mice treated with LPS/elastase were stimulated with bone marrow–derived dendritic cells (BMDCs)
alone (media), or BMDCs pulsed with collagen or elastin. (K) CD41 T cells were sorted by flow cytometry from pooled lung and airway samples of
five mice treated with LPS/elastase. Sorted CD41 T cells were then stimulated with BMDCs alone (media) or BMDCs pulsed with collagen or elastin.
*P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. PBS = phosphate-buffered saline; PD-1 = programmed cell death protein 1.

980 American Journal of Respiratory and Critical Care Medicine Volume 193 Number 9 | May 1 2016
ORIGINAL ARTICLE

A chao1 (Richness) Shannon (Diversity)


2000 6.3

6.2

6.1
1500
6.0

Shannon
5.9
chao1

1000
5.8

PBS (4 pooled BALF) 5.7 PBS (4 pooled BALF)


PBS (4 pooled BALF) PBS (4 pooled BALF)
500 5.6
LPS/Elastase (4 pooled BALF) LPS/Elastase (4 pooled BALF)
LPS/Elastase (4 pooled BALF) 5.5 LPS/Elastase (4 pooled BALF)
Negative control 5 Negative control
0 0

00

10 0
14 0
18 0
22 0
26 0
30 0
34 0
38 0
42 0
46 0
50 0
54 0
58 0
62 0
66 0
70 0
74 0
78 0
82 0
86 0
90 0
94 0
98 0
0
00

10 0
14 0
18 0
22 0
26 0
30 0
34 0
38 0
42 0
46 0
50 0
54 0
58 0
62 0
66 0
70 0
74 0
78 0
82 0
86 0
90 0
94 0
98 0
0

0
00
00
00
00
00
00
00
00
00
00
00
00
00
00
00
00
00
00
00
00
00
00
00
0
00
00
00
00
00
00
00
00
00
00
00
00
00
00
00
00
00
00
00
00
00
00
00

20
60
20
60

N seqs/sample N seqs/sample

B hclust (ward.D), Bray-Curtis distance Relative abundance %

0 10 20 30
k__Bacteria;p__Actinobacteria;c__Actinobacteria;o__Actinomycetales;f__;g__
k__Bacteria;p__Actinobacteria;c__Actinobacteria;o__Actinomycetales;f__ACK−M1;g__
k__Bacteria;p__Actinobacteria;c__Actinobacteria;o__Actinomycetales;f__Actinomycetaceae;g__N09
k__Bacteria;p__Actinobacteria;c__Actinobacteria;o__Actinomycetales;f__Corynebacteriaceae;g__Corynebacterium
k__Bacteria;p__Actinobacteria;c__Actinobacteria;o__Actinomycetales;f__Geodermatophilaceae;g__
k__Bacteria;p__Actinobacteria;c__Actinobacteria;o__Actinomycetales;f__Microbacteriaceae;g__
k__Bacteria;p__Actinobacteria;c__Actinobacteria;o__Actinomycetales;f__Microbacteriaceae;g__Candidatus Rhodoluna
k__Bacteria;p__Actinobacteria;c__Actinobacteria;o__Actinomycetales;f__Microbacteriaceae;g__Clavibacter
k__Bacteria;p__Actinobacteria;c__Actinobacteria;o__Actinomycetales;f__Microbacteriaceae;g__Microbacterium
k__Bacteria;p__Actinobacteria;c__Actinobacteria;o__Actinomycetales;f__Micrococcaceae;Other
k__Bacteria;p__Actinobacteria;c__Actinobacteria;o__Actinomycetales;f__Micrococcaceae;g__Micrococcus
k__Bacteria;p__Actinobacteria;c__Actinobacteria;o__Actinomycetales;f__Propionibacteriaceae;g__Propionibacterium
k__Bacteria;p__Bacteroidetes;c__Bacteroidia;o__Bacteroidales;f__Porphyromonadaceae;g__Paludibacter
k__Bacteria;p__Bacteroidetes;c__Bacteroidia;o__Bacteroidales;f__Prevotellaceae;g__
k__Bacteria;p__Bacteroidetes;c__Bacteroidia;o__Bacteroidales;f__Prevotellaceae;g__Prevotella
k__Bacteria;p__Bacteroidetes;c__Bacteroidia;o__Bacteroidales;f__Rikenellaceae;g__
k__Bacteria;p__Bacteroidetes;c__Bacteroidia;o__Bacteroidales;f__S24−7;g__
k__Bacteria;p__Bacteroidetes;c__Bacteroidia;o__Bacteroidales;f__[Odoribacteraceae];g__Odoribacter
k__Bacteria;p__Bacteroidetes;c__Cytophagia;o__Cytophagales;f__Cytophagaceae;g__
k__Bacteria;p__Bacteroidetes;c__Flavobacteriia;o__Flavobacteriales;f__Cryomorphaceae;g__
k__Bacteria;p__Bacteroidetes;c__Flavobacteriia;o__Flavobacteriales;f__Cryomorphaceae;g__Fluviicola
k__Bacteria;p__Bacteroidetes;c__Flavobacteriia;o__Flavobacteriales;f__Flavobacteriaceae;g__Flavobacterium
k__Bacteria;p__Bacteroidetes;c__Flavobacteriia;o__Flavobacteriales;f__[Weeksellaceae];Other
k__Bacteria;p__Bacteroidetes;c__Flavobacteriia;o__Flavobacteriales;f__[Weeksellaceae];g__Chryseobacterium
k__Bacteria;p__Bacteroidetes;c__Flavobacteriia;o__Flavobacteriales;f__[Weeksellaceae];g__Cloacibacterium
k__Bacteria;p__Bacteroidetes;c__[Saprospirae];o__[Saprospirales];f__Chitinophagaceae;g__Niabella
k__Bacteria;p__Cyanobacteria;c__Chloroplast;o__Cryptophyta;f__;g__
k__Bacteria;p__Cyanobacteria;c__Chloroplast;o__Stramenopiles;f__;g__
k__Bacteria;p__Cyanobacteria;c__Chloroplast;o__Streptophyta;f__;g__
k__Bacteria;p__Deferribacteres;c__Deferribacteres;o__Deferribacterales;f__Deferribacteraceae;g__Mucispirillum
k__Bacteria;p__Firmicutes;c__Bacilli;o__Bacillales;f__Staphylococcaceae;g__Staphylococcus
k__Bacteria;p__Firmicutes;c__Bacilli;o__Lactobacillales;f__Lactobacillaceae;g__Lactobacillus
k__Bacteria;p__Firmicutes;c__Bacilli;o__Lactobacillales;f__Streptococcaceae;g__Streptococcus
k__Bacteria;p__Firmicutes;c__Bacilli;o__Turicibacterales;f__Turicibacteraceae;g__Turicibacter
k__Bacteria;p__Firmicutes;c__Clostridia;o__Clostridiales;f__;g__
k__Bacteria;p__Firmicutes;c__Clostridia;o__Clostridiales;f__Ruminococcaceae;g__Oscillospira
k__Bacteria;p__Firmicutes;c__Erysipelotrichi;o__Erysipelotrichales;f__Erysipelotrichaceae;g__[Eubacterium]
k__Bacteria;p__GN02;c__3BR−5F;o__;f__;g__
k__Bacteria;p__GN02;c__GKS2−174;o__;f__;g__
k__Bacteria;p__OD1;c__SM2F11;o__;f__;g__
k__Bacteria;p__OD1;c__ZB2;o__;f__;g__
k__Bacteria;p__Proteobacteria;c__Alphaproteobacteria;o__;f__;g__
k__Bacteria;p__Proteobacteria;c__Alphaproteobacteria;o__Caulobacterales;f__Caulobacteraceae;g__
k__Bacteria;p__Proteobacteria;c__Alphaproteobacteria;o__Caulobacterales;f__Caulobacteraceae;g__Caulobacter
k__Bacteria;p__Proteobacteria;c__Alphaproteobacteria;o__Rhizobiales;f__Hyphomicrobiaceae;g__Hyphomicrobium
k__Bacteria;p__Proteobacteria;c__Alphaproteobacteria;o__Rhizobiales;f__Methylobacteriaceae;g__Methylobacterium
k__Bacteria;p__Proteobacteria;c__Alphaproteobacteria;o__Rhizobiales;f__Phyllobacteriaceae;Other
k__Bacteria;p__Proteobacteria;c__Alphaproteobacteria;o__Rhodobacterales;f__Rhodobacteraceae;g__Rhodobacter
k__Bacteria;p__Proteobacteria;c__Alphaproteobacteria;o__Rickettsiales;f__Rickettsiaceae;g__
k__Bacteria;p__Proteobacteria;c__Alphaproteobacteria;o__Sphingomonadales;f__;g__
k__Bacteria;p__Proteobacteria;c__Alphaproteobacteria;o__Sphingomonadales;f__Erythrobacteraceae;g__
k__Bacteria;p__Proteobacteria;c__Alphaproteobacteria;o__Sphingomonadales;f__Sphingomonadaceae;g__
k__Bacteria;p__Proteobacteria;c__Betaproteobacteria;o__;f__;g__
k__Bacteria;p__Proteobacteria;c__Betaproteobacteria;o__Burkholderiales;f__Burkholderiaceae;g__Burkholderia
k__Bacteria;p__Proteobacteria;c__Betaproteobacteria;o__Burkholderiales;f__Comamonadaceae;Other
k__Bacteria;p__Proteobacteria;c__Betaproteobacteria;o__Burkholderiales;f__Comamonadaceae;g__
k__Bacteria;p__Proteobacteria;c__Betaproteobacteria;o__Burkholderiales;f__Comamonadaceae;g__Delftia
k__Bacteria;p__Proteobacteria;c__Betaproteobacteria;o__Burkholderiales;f__Oxalobacteraceae;g__
k__Bacteria;p__Proteobacteria;c__Betaproteobacteria;o__Burkholderiales;f__Oxalobacteraceae;g__Cupriavidus
k__Bacteria;p__Proteobacteria;c__Betaproteobacteria;o__Burkholderiales;f__Oxalobacteraceae;g__Polynucleobacter
k__Bacteria;p__Proteobacteria;c__Betaproteobacteria;o__Procabacteriales;f__Procabacteriaceae;g__
k__Bacteria;p__Proteobacteria;c__Deltaproteobacteria;o__Bdellovibrionales;f__Bdellovibrionaceae;g__Bdellovibrio
k__Bacteria;p__Proteobacteria;c__Deltaproteobacteria;o__MIZ46;f__;g__
k__Bacteria;p__Proteobacteria;c__Deltaproteobacteria;o__Myxococcales;f__;g__
k__Bacteria;p__Proteobacteria;c__Deltaproteobacteria;o__Spirobacillales;f__;g__
k__Bacteria;p__Proteobacteria;c__Epsilonproteobacteria;o__Campylobacterales;f__Campylobacteraceae;g__Sulfurospirillum
k__Bacteria;p__Proteobacteria;c__Gammaproteobacteria;o__Alteromonadales;f__Alteromonadaceae;g__Cellvibrio
k__Bacteria;p__Proteobacteria;c__Gammaproteobacteria;o__Alteromonadales;f__[Chromatiaceae];g__Rheinheimera
k__Bacteria;p__Proteobacteria;c__Gammaproteobacteria;o__Legionellales;f__Legionellaceae;g__Legionella
k__Bacteria;p__Proteobacteria;c__Gammaproteobacteria;o__Pseudomonadales;f__Moraxellaceae;g__Acinetobacter
k__Bacteria;p__Proteobacteria;c__Gammaproteobacteria;o__Pseudomonadales;f__Pseudomonadaceae;g__Pseudomonas
k__Bacteria;p__SR1;c__;o__;f__;g__
k__Bacteria;p__TM7;c__;o__;f__;g__
k__Bacteria;p__TM7;c__TM7−1;o__;f__;g__
k__Bacteria;p__TM7;c__TM7−3;o__EW055;f__;g__
k__Bacteria;p__[Thermi];c__Deinococci;o__Deinococcales;f__Deinococcaceae;g__CM44
Negative LPS/Elastase PBS
control

C g_Prevotella g_Pseudomonas g_Lactobacillus g_Chryseobacterium


Relative abundance (%)

0.003 0.025 0.08 0.020

0.020
0.06 0.015
0.002
0.015
0.04 0.010
0.010
0.001
0.02 0.005
0.005

0.000 0.000 0.00 0.000


PBS LPS/Elastase PBS LPS/Elastase PBS LPS/Elastase PBS LPS/Elastase

Figure 3. The pulmonary microbiota is altered upon the induction of chronic pulmonary inflammation. Eight mice per group were treated with phosphate-
buffered saline (PBS) or LPS/elastase for 4 weeks, and DNA was extracted from four pooled bronchoalveolar lavage fluid (BALF) samples per group,
resulting in two sample sets per group for Illumina MiSeq 16S rDNA sequencing. (A) Richness (chao1) and diversity (Shannon) rarefaction curves of the
16S rDNA library constructed from pooled BALF of PBS- or LPS/elastase–treated mice. (B) Heat map showing the relative abundance of major bacterial
genera (operational taxonomic units . 0.05% of relative abundance in minimum two samples) in pooled BALF of PBS- or LPS/elastase–treated mice and
negative control. Dendrogram was drawn from Ward hierarchical clustering algorithm using Bray-Curtis dissimilarity matrix. (C) Bar graphs representing
the relative abundance of specific genera showing differences between the BALF of PBS- and LPS/elastase–treated mice.

Yadava, Pattaroni, Sichelstiel, et al.: Microbiota Sustains Chronic Pulmonary Inflammation 981
ORIGINAL ARTICLE

A B C
Water Antibiotic
110 0.09
FEV0.1/FVC(%)

(mL/cmH2O)
100

Compliance
0.08
90
0.07
80
70 0.06
60
Water Antibiotic Water Antibiotic 100 m 100 m

D ** E Water Antibiotic **
1.5 2.0

Neutrophils in BAL
Total cells in BAL

1.2 1.5
(X106)

(X105)
0.9
1.0
0.6
Ly6g

0.3 0.5

Water Antibiotic Ly6C Water Antibiotic

F Water Antibiotic
4 **
4
aM in BAL(X105)

iM in BAL(X104)
3 3
2 2
CD11c

1 1

F480 Water Antibiotic Water Antibiotic

G H Total antibodies in BAL I Specific IgG1 J Specific IgA


25 * Water 50
** 0.04 0.04 *
20 Antibiotic 40 ** **
No./section

0.03 0.03
of the lung

g/ml
g/ml

g/ml

15 30
0.02 0.02
10 20
5 10 0.01 0.01

VALT BALT ALT IgG1 IgA IgM Collagen Elastin Collagen Elastin
Figure 4. Microbiota-derived signals amplify innate and adaptive inflammation in disease. Mice harboring a specific pathogen–free microflora (water)
or mice depleted of microbiota by antibiotic treatment were subjected to LPS/elastase treatment over 4 weeks. Analysis was performed a week after last
challenge. (A) Lung compliance and (B) FEV/FVC were measured using FlexiVent invasive airway mechanics system. Data are pooled from two
experiments. (C) Representative slides of hematoxylin and eosin–stained lung sections showing peribronchiolar and perivascular inflammation in water-
and antibiotic-treated groups. (D) Number of cells infiltrating the bronchoalveolar lavage (BAL) was determined. (E) Neutrophils and (F) alveolar (aMF) and
interstitial (iMF) macrophages in the BAL were enumerated by flow cytometry. Data are representative of two independent experiments. (G) Vascular-
associated lymphoid tissues (VALT), alveolar-associated lymphoid tissues (ALT), or bronchial-associated lymphoid tissues (BALT) were quantified in
hematoxylin and eosin–stained sections of lungs. Error bars represent SEM (n = 4). (H) Levels of IgG1, IgA, and IgM in BAL supernatant were measured by
ELISA. Collagen- and elastin-specific (I) IgG1 and (J) IgA responses were quantified by ELISA. Error bars represent SEM. Data pooled from two
experiments (n = 9). *P < 0.05, **P < 0.01.

similar to COPD in humans. We were able T cells and CD41 T cells increasingly (Figure 6F). We found that neutralizing
to distinguish mild and severe disease based accumulated in the lungs with repeated IL-17A did not improve FEV0.1/FVC but
on the decline in lung function seen at LPS/elastase challenges (P , 0.01) (Figures reduced lung compliance significantly
different time points in the disease model 6D and 6E, Figure E7). Based on the (P , 0.05) (Figures 6G and 6H) in addition
(Figures 6A and 6B). We found that the kinetics observed in mild versus severe to the total cellular and neutrophilic
kinetics of IL-17A production followed disease, we performed a targeted infiltration into the BAL (P , 0.01)
the progression of disease (Figure 6C). neutralization of IL-17A starting 1 day (Figures 6I and 6J). Neutralizing IL-17A
Moreover, both IL-17–producing gd1 before the second LPS/elastase challenge also reduced the total frequency of

982 American Journal of Respiratory and Critical Care Medicine Volume 193 Number 9 | May 1 2016
ORIGINAL ARTICLE

A B

IL-17A+ cells (X106)


* Sources of IL-17A
Gated on live cells 5.0
Water Antibiotic T cells
Water Antibiotic 4.0
3.98% 5.06% CD3- cells
3.0 14.33% 26.69 15.88%
31.05 CD3+CD4–,
2.0 % 0.56% % 0.57% CD8–,– cells
IL-17A

1.0 CD8+ T cells


50.09% 51.81% CD4+ T cells
IFN Water Antibiotic

C ** D
Gated on CD4+ T cells Gated on + T cells 2.0
1.5

T cells i(X106)
IL-17A+ CD4+
T cells (X106)

IL-17A+ +
Water Antibiotic Water Antibiotic 1.5
1.0
1.0
0.5
IL-17A

IL-17A
0.5

IFN Water Antibiotic IFN Water Antibiotic

E F G
Gated on live cells 1.5 * 1.5 1.5
*

T cells i(×105)
IL-17A+ CD4+

PBS LPS/Elastase

IL-17A+ +
cells (X106)
cells (X106)

microbiome Microbiome 1.0 1.0 1.0


IL-17A+

0.5 0.5 0.5


IL-17A

PBS LPS/elastase PBS LPS/elastase PBS LPS/elastase


IFN
Source of microbiome Source of microbiome Source of microbiome
Figure 5. Microbiota enhances production of IL-17A by CD41 and gd1 T cells. Cells isolated from lungs and airways of control (water) or antibiotic were
stimulated in vitro with phorbol myristate acetate, ionomycin, and monensin for 4 hours. (A) Ex vivo stimulated cells were surface stained, fixed, and
permeabilized for detection of cytokines IFN-g and IL-17A. (B) Cellular sources of IL-17A were analyzed by flow cytometry. Pie charts represent the
composition based on mean values from four to five mice per group. (C) Total number of CD41 T cells producing IL-17A or (D) gd1 T cells was determined
by flow cytometry. Data are representative of two independent experiments. (E) Antibiotic-treated mice were challenged with LPS/elastase on Day 0, 7,
and 21. Every day starting at Day 12 until Day 20 and then on Days 22 to 24, they received microbiota (sequenced in Figure 3) isolated from either
phosphate-buffered saline (PBS)-challenged specific pathogen–free (SPF) mice or LPS/elastase-challenged SPF mice in 100 ml intranasally. On Day 25,
cells isolated from the lungs of the recolonized antibiotic treated recipients were restimulated with phorbol myristate acetate ionomycin, and IL-17
production was analyzed by flow cytometry. Total number of IL-17A–producing (F) CD41 T cells and (G) gd1 T cells were also determined by flow
cytometry. *P < 0.05, **P < 0.01.

lymphoid follicles (P , 0.05) without such as inflammatory bowel disease (19, 20). changes seen in microbial composition
specifically affecting any particular subtype Although the gut microbiota can impact in the airways of mice as in humans.
(VALT, alveolar-associated lymphoid tissue, systemic immune responses, there is Specifically, we found that the overall
or bronchiolar-associated lymphoid tissue) an emerging paradigm supporting the diversity of the pulmonary microbiome
(Figure 6K). This was also associated with a importance of host–microbe interactions was reduced, Prevotella was absent, and
reduction in total IgG1 (P , 0.05), whereas localized within a single habitat, such as the bacterial genera Pseudomonas and
IgA and IgM were increased (P , 0.05, that found in the lung and skin (55, 56). Lactobacillus were increased in abundance
P , 0.01, respectively) (Figure 6L). Overall, Several studies have shown that the in the LPS/elastase–treated animals as
targeting the IL-17A pathway was effective at composition of the airway microbiota in compared with control animals (43). This
ameliorating inflammation and uncoupling healthy lungs differs from lungs of patients correlates with studies in humans showing
the autoimmune component of the disease. with COPD, although as yet whether that Prevotella is a common colonizer of the
disease creates a habitat for the bacteria or airways of healthy subjects when compared
whether the bacteria cause disease hasn’t with patients with asthma or COPD (13,
Discussion been delineated (10, 13, 14). Our findings 43). Consistently Pseudomonas is increased
implicate a functional role for the in patients with asthma and COPD (43),
Alterations in the gut microbiome have been microbiota in COPD. It is noteworthy that and its presence correlates with COPD
extensively documented for chronic diseases there were many similarities in the types of severity (44). Similarly, the relative

Yadava, Pattaroni, Sichelstiel, et al.: Microbiota Sustains Chronic Pulmonary Inflammation 983
ORIGINAL ARTICLE

0.09 100

FEV 0.1/FVC (%)


A PBS or LPS/Elastase B * PBS

(cm H2O.s/ml)
**

Compliance
0.08 LPS/Elastase
80
0.07
0.06 60
Week 1 2 3 4 5 0.05
1 2 3 4 1 2 3 4
Analysis Analysis Analysis Analysis No. of LPS/Elastase No. of LPS/Elastase
challenges challenges

C D E

IL-17A+ CD4+ cells


No. of LPS/Elastase challenges 107 106

IL-17A+  cells


** 106 ** **
** **

IL-17A+ cells
**
1X 2X 3X 4X 106 10 5
**
* 10 5
* ***
IL-17A

105 * ** *
104 104

RORt 1 2 3 4 1 2 3 4 1 2 3 4
No. of LPS/Elastase No. of LPS/Elastase No. of LPS/Elastase
challenges challenges challenges

F PBS or LPS/Elastase G H *
Analysis
100 0.065
FEV0.1/FVC (%)

(mL/cmH2O)
Compliance
80
0.060
60
0 6 7 10 13 14 17 20 21 24 28 0.055
40
20 0.050

IL-17A or isotype control antibody treatment Isotype IL-17A Isotype IL-17A


I J K L
* * * * Isotype
Cells in BAL (X105)

30
Neutrophils (X105)

No./section of lung

4.5 1.5 50 IL-17A


4.0 g/ml
40
1.0 20
3.5 30 *
3.0 10 20
0.5 **
2.5 10
Isotype IL-17A Isotype IL-17A Isotype IL-17A IgG1 IgA IgM
Figure 6. IL-17A kinetics correlate with disease severity, and targeted neutralization of IL-17A ameliorates disease. (A) BALB/c mice were challenged with
one, two, three, or four doses of LPS/elastase. Analysis was performed 1 week after the indicated number of challenges. (B) Invasive lung function
measurement was performed using the FlexiVent system. (C) The kinetics of RORgt-expressing and IL-17A–producing ex vivo stimulated cells from lungs
and airways of mice challenged with one, two, three, or four times phosphate-buffered saline (PBS) or LPS/elastase (n = 4–5 per time point per group).
The kinetics of IL-17A–producing (D) CD41 and (E) gd1 T cells in mice challenged with one, two, three, or four times PBS or LPS/elastase (n = 4–5 per time
point per group). (F) Targeted neutralization of IL-17A based on its production in mild to severe disease. Mice were challenged with LPS/elastase
over 4 weeks and administered anti–IL-17A antibody or isotype control at indicated time points. (G) Lung compliance and (H) FEV/FVC were measured
using the FlexiVent invasive airway mechanics system. (I) Number of cells recovered in the bronchoalveolar lavage (BAL) was determined. (J) Neutrophils
in the BAL were enumerated by differential cell counts. Data are representative of at least two independent experiments. (K) Vascular-associated,
alveolar-associated, or bronchial-associated lymphoid follicles were quantified in hematoxylin and eosin sections of lungs. Error bars represent SEM
(n = 4–5). (L) Levels of IgG1, IgA, and IgM in BAL supernatant were measured by ELISA. Data pooled from two experiments (n = 9). *P < 0.05, **P < 0.01,
***P < 0.001. RORgt = retinoid-related orphan receptor gt.

abundance of Lactobacillus is also increased A key question still facing the field is pressure facilitating colonization by specific
in patients with asthma and COPD (43), where does the airway microbiota originate? bacteria. Considering our experiments were
and an additional study has linked it to (57). Recent studies suggest the pulmonary performed under controlled conditions
COPD severity (10). Chryseobacterium microbiome originates from the dispersal of with mice housed in isolated and filtered
levels were also increased in the BAL of microbes from the nasal and oral cavities cages, our results support the concept
LPS/elastase–treated mice. This did not (58, 59). It is possible that this “seeding” that inflammatory conditions allow the
correlate with any published data in population is altered in patients with outgrowth of bacteria that are part of the
patients with COPD, but studies have COPD, and this subsequently affects the steady-state “healthy” microbiota.
shown that Chryseobacterium infection in microbiome in the lungs. Alternatively, Increased levels of IL-17A have been
individuals with cystic fibrosis correlates the state of chronic inflammation in the implicated in stable COPD (52), and a
with Pseudomonas (45). diseased lungs may provide a selection recent study has shown that the number of

984 American Journal of Respiratory and Critical Care Medicine Volume 193 Number 9 | May 1 2016
ORIGINAL ARTICLE

CD41 IL-17A1 cells in the small airways bacterial biomass in the airways of mice, we LFs and result in the production of
positively correlates with airflow limitation are limited with feasible approaches to autoantibodies (22, 64). Although sequence
(46). Tracking the development of disease address this point. As such, from this study analysis of B cell clones isolated from LFs in
progression in our model showed that we can only conclude the microbiota COPD lungs has indicated the presence of
IL-17A production increased with disease changes in the airways of mice treated with an oligoclonal, antigen-specific humoral
severity. Moreover, we observed temporal LPS/elastase, similar to that reported in response (25), there are conflicting reports
changes in the cellular sources of IL-17A humans; depletion (antibiotics) or absence regarding the production of self-reactive
(Figure E7). We found that gd1 T cells (axenic/germ-free mice) of a microbiota antibodies (28–32). It is important to note
accounted for the majority of the few ameliorates disease; and transfer of a that most of these studies assess systemic
IL-17A–producing cells in the healthy and microbially enriched fraction of the BAL antibody levels, which may not be
mildly diseased lungs. In severe disease from diseased animals increases the disease- representative of responses within the lung.
there was an expansion of both causing IL-17 inflammatory pathway in Another caveat in these studies is the
IL-17A–producing CD41 and gd1 T cells. recipient mice. A further possibility is that identification of the relevant autoantigen,
Signals from commensal bacteria have been the role of the microbiota in promoting which may vary considerably among
shown to enhance the expansion of both disease in our model is due to the gut patients. In our model and in the cigarette
gd1 T cells and Th17 cells (34, 60–62) and microbiota; indeed, we have previously smoke–induced model of emphysema, LF
exacerbate IL-17–mediated inflammation reported a gut–lung axis, with mice fed a formation is associated with an increase in
in experimental autoimmune encephalitis high-fiber diet being protected against local antibody levels in the BAL, whereas
and arthritis (34–36). In particular, specific allergic airway inflammation (63). systemic antibodies were unaffected (25,
bacteria can induce Th17 responses, among However, in the current study we expect 26). In line with these data, the deposition
which segmented filamentous bacteria have the airway microbiota to play a more of IgG complexes has been demonstrated
been extensively studied (34, 36). Although fundamental role, as we did not find in lungs of patients with severe COPD,
segmented filamentous bacteria were not changes in the gut microbiota in our model although their antigenic specificity is
detected in the lung in our study, it is likely (data not shown). It remains possible that unknown (30, 65). A limitation of our study
that the outgrowth of other lung bacteria certain changes in the gut microbiota could is that we investigated self-reactive
resident in the diseased lungs could also impact the development of COPD, and antibodies to only two self-antigens,
promote IL-17 responses. future in-depth metagenomic approaches collagen and elastin, which constitute only
In the current study, we attempted to could provide the answer. a portion of the breakdown products of the
transfer the microbiota from the BAL of Concurrent with the development of lung tissue. In the future it would be
either PBS- or LPS/elastase–treated mice the IL-17 response in our model, we valuable to comprehensively characterize
into the airways of mice depleted (with found an autoimmune component of the the repertoire of the self-reactive antibodies
antibiotics) or devoid (germ-free) of a inflammation evolved. It is tempting to in patients with COPD and investigate
microbiota. This transfer consisted of a speculate that the microbially enhanced whether specific antibody signatures are
microbial pellet that had been isolated IL-17 response promotes an autoimmune correlated with disease severity. This could
(excluding host cells and soluble component, which further perpetuates distinguish a subset of patients with COPD
components in suspension) from the BAL the disease. The increased availability of in whom disease pathogenesis has a clear
performed on mice; however, in addition to self-antigens due to the destruction of lung association with autoimmunity and
the enriched microbial components of this tissue might also allow the expansion of ultimately identify patients who would
pellet, acellular material from the lungs autoreactive T cells. Further supporting benefit the most from treatments targeting
could also have been enriched. Transfer of an autoimmune component to disease B cells (66, 67).
the microbiota-enriched solution derived pathogenesis, lymphoid follicles have The beneficial versus detrimental role
from mice that had previously been treated consistently been observed in animal of local antibody responses could be
with LPS/elastase increased the production models and in humans with COPD determined by different antigen specificities
of IL-17 in recipient mice, as compared (24–27). Recently, Bracke and colleagues and associated isotypes. It has been reported
with the solution derived from mice demonstrated a pathogenic role of LFs in that compared with patients with COPD,
previously treated with PBS. Whether this disease (26). In this study they abrogated healthy smokers exhibit a preferential
experimental approach led to a sufficiently LF formation by targeting CXCL13 and switching to IgA (68). IgA responses at
large increase in IL-17 levels to influence observed a decrease in the local antibody mucosal surfaces are associated with
lung function remains to be determined. response with a concomitant improvement protective immunity, and blunted IgA
There are several technical limitations in specific aspects of disease such as responses in COPD can be linked to
to the experimental approach of transferring inflammation and alveolar wall destruction. increased susceptibility to infections (69).
the airway microbiota. For instance, it is Microbial colonization and infections are Conversely, self-reactive antibodies, in
unclear whether viable bacteria are required more frequent in patients with very severe particular IgG1 isotypes, are more prevalent
to see this effect. Another important caveat disease. From an evolutionary standpoint, in severe disease and implicated in
is we are unable to distinguish between the LFs in chronic lung diseases might arise to pathogenesis (30, 65, 68). In our study,
influence of the microbes and that of other combat infections by enhancing the local depleting the microbiota had a profound
potential constituents of the processed immune response. However, self-antigens effect on the total IgA levels, suggesting that
BALF, for example, cellular debris. With the derived from the collateral damage of the a proportion of these antibody responses
current state of the art and the very low lung tissue might also be presented within may be directed against the microbiota.

Yadava, Pattaroni, Sichelstiel, et al.: Microbiota Sustains Chronic Pulmonary Inflammation 985
ORIGINAL ARTICLE

However, the absence of a microbiota also Our data implicate the airway pathogenic mechanism linked with
reduced the levels of self-reactive IgA and microbiota as a key player in the promotion COPD. The characterization of dysbiotic
IgG1 antibodies. Whether this is due to the of local IL-17A responses and, consequently, microbial communities and their
cross-reactivity between microbial and the development of chronic lung disease. associated inflammatory signature could
self-antigens or an indirect effect by the This pathway impacts both innate and pave the way for personalized medicine
induction of IL-17, as seen in a model of adaptive inflammatory processes and to disrupt the chronic cycle of tissue
autoimmune arthritis (34), is unknown. regulates lymphoid neogenesis and the damage and the heterogeneity seen in
Our data suggest that microbial cues could expansion of autoreactive T and B cells. The COPD. n
impact not only the specificity but also the microbiota-enhanced local autoimmunity
isotype of antibody responses generated likely contributes to disease chronicity, Author disclosures are available with the text
locally. although autoimmunity only represents one of this article at www.atsjournals.org.

References 19. Frank DN, St Amand AL, Feldman RA, Boedeker EC, Harpaz N, Pace
NR. Molecular-phylogenetic characterization of microbial community
1. Rosenbaum L, Lamas D. Facing a “slow-motion disaster”: the UN imbalances in human inflammatory bowel diseases. Proc Natl Acad
meeting on noncommunicable diseases. N Engl J Med 2011;365: Sci USA 2007;104:13780–13785.
2345–2348. 20. Manichanh C, Rigottier-Gois L, Bonnaud E, Gloux K, Pelletier E,
2. Halbert RJ, Natoli JL, Gano A, Badamgarav E, Buist AS, Mannino DM. Frangeul L, Nalin R, Jarrin C, Chardon P, Marteau P, et al. Reduced
Global burden of COPD: systematic review and meta-analysis. diversity of faecal microbiota in Crohn’s disease revealed by a
Eur Respir J 2006;28:523–532. metagenomic approach. Gut 2006;55:205–211.
3. Fletcher C, Peto R. The natural history of chronic airflow obstruction. 21. Agustı́ A, MacNee W, Donaldson K, Cosio M. Hypothesis: does COPD
BMJ 1977;1:1645–1648. have an autoimmune component? Thorax 2003;58:832–834.
4. Saetta M, Finkelstein R, Cosio MG. Morphological and cellular basis for 22. Brusselle GG, Demoor T, Bracke KR, Brandsma CA, Timens W.
airflow limitation in smokers. Eur Respir J 1994;7:1505–1515. Lymphoid follicles in (very) severe COPD: beneficial or harmful? Eur
5. Tuder RM, Petrache I. Pathogenesis of chronic obstructive pulmonary Respir J 2009;34:219–230.
disease. J Clin Invest 2012;122:2749–2755. 23. Aloisi F, Pujol-Borrell R. Lymphoid neogenesis in chronic inflammatory
6. Finkelstein R, Fraser RS, Ghezzo H, Cosio MG. Alveolar inflammation diseases. Nat Rev Immunol 2006;6:205–217.
and its relation to emphysema in smokers. Am J Respir Crit Care Med 24. Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L,
1995;152:1666–1672. Cherniack RM, Rogers RM, Sciurba FC, Coxson HO, et al. The
7. Barnes PJ. Mediators of chronic obstructive pulmonary disease. nature of small-airway obstruction in chronic obstructive pulmonary
Pharmacol Rev 2004;56:515–548. disease. N Engl J Med 2004;350:2645–2653.
8. Willemse BW, ten Hacken NH, Rutgers B, Lesman-Leegte IG, Postma 25. van der Strate BW, Postma DS, Brandsma CA, Melgert BN, Luinge MA,
DS, Timens W. Effect of 1-year smoking cessation on airway Geerlings M, Hylkema MN, van den Berg A, Timens W, Kerstjens HA.
inflammation in COPD and asymptomatic smokers. Eur Respir J 2005; Cigarette smoke-induced emphysema: a role for the B cell? Am J
26:835–845. Respir Crit Care Med 2006;173:751–758.
9. Zakharkina T, Heinzel E, Koczulla RA, Greulich T, Rentz K, Pauling JK, 26. Bracke KR, Verhamme FM, Seys LJ, Bantsimba-Malanda C,
Baumbach J, Herrmann M, Grünewald C, Dienemann H, et al. Analysis Cunoosamy DM, Herbst R, Hammad H, Lambrecht BN, Joos GF,
of the airway microbiota of healthy individuals and patients with Brusselle GG. Role of CXCL13 in cigarette smoke-induced lymphoid
chronic obstructive pulmonary disease by T-RFLP and clone follicle formation and chronic obstructive pulmonary disease. Am J
sequencing. Plos One 2013;8:e68302. Respir Crit Care Med 2013;188:343–355.
10. Sze MA, Dimitriu PA, Hayashi S, Elliott WM, McDonough JE, Gosselink 27. Mori M, Andersson CK, Svedberg KA, Glader P, Bergqvist A,
JV, Cooper J, Sin DD, Mohn WW, Hogg JC. The lung tissue Shikhagaie M, Löfdahl CG, Erjefält JS. Appearance of remodelled
microbiome in chronic obstructive pulmonary disease. Am J Respir and dendritic cell-rich alveolar-lymphoid interfaces provides
Crit Care Med 2012;185:1073–1080. a structural basis for increased alveolar antigen uptake in
11. Han MK, Huang YJ, Lipuma JJ, Boushey HA, Boucher RC, Cookson chronic obstructive pulmonary disease. Thorax 2013;68:
WO, Curtis JL, Erb-Downward J, Lynch SV, Sethi S, et al. 521–531.
Significance of the microbiome in obstructive lung disease. Thorax 28. Bonarius HP, Brandsma CA, Kerstjens HA, Koerts JA, Kerkhof M,
2012;67:456–463. Nizankowska-Mogilnicka E, Roozendaal C, Postma DS, Timens W.
12. Erb-Downward JR, Huffnagle GB, Martinez FJ. The microbiota in Antinuclear autoantibodies are more prevalent in COPD in
respiratory disease. Am J Respir Crit Care Med 2012;185:1037–1038. association with low body mass index but not with smoking history.
13. Erb-Downward JR, Thompson DL, Han MK, Freeman CM, McCloskey Thorax 2011;66:101–107.
L, Schmidt LA, Young VB, Toews GB, Curtis JL, Sundaram B, et al. 29. Cottin V, Fabien N, Khouatra C, Moreira A, Cordier JF. Anti-elastin
Analysis of the lung microbiome in the “healthy” smoker and in autoantibodies are not present in combined pulmonary fibrosis and
COPD. Plos One 2011;6:e16384. emphysema. Eur Respir J 2009;33:219–221.
14. Hilty M, Burke C, Pedro H, Cardenas P, Bush A, Bossley C, Davies J, 30. Feghali-Bostwick CA, Gadgil AS, Otterbein LE, Pilewski JM, Stoner
Ervine A, Poulter L, Pachter L, et al. Disordered microbial MW, Csizmadia E, Zhang Y, Sciurba FC, Duncan SR. Autoantibodies
communities in asthmatic airways. Plos One 2010;5:e8578. in patients with chronic obstructive pulmonary disease. Am J Respir
15. Chambers DC, Gellatly SL, Hugenholtz P, Hansbro PM. JTD special Crit Care Med 2008;177:156–163.
edition ’Hot Topics in COPD’: the microbiome in COPD. J Thorac Dis 31. Lee SH, Goswami S, Grudo A, Song LZ, Bandi V, Goodnight-White S,
2014;6:1525–1531. Green L, Hacken-Bitar J, Huh J, Bakaeen F, et al. Antielastin
16. Cui L, Morris A, Huang L, Beck JM, Twigg HL III, von Mutius E, Ghedin E. autoimmunity in tobacco smoking-induced emphysema. Nat Med
The microbiome and the lung. Ann Am Thorac Soc 2014;11:S227–S232. 2007;13:567–569.
17. Dickson RP, Martinez FJ, Huffnagle GB. The role of the microbiome in 32. Rinaldi M, Lehouck A, Heulens N, Lavend’homme R, Carlier V,
exacerbations of chronic lung diseases. Lancet 2014;384:691–702. Saint-Remy JM, Decramer M, Gayan-Ramirez G, Janssens W.
18. Rogers GB, Shaw D, Marsh RL, Carroll MP, Serisier DJ, Bruce KD. Antielastin B-cell and T-cell immunity in patients with
Respiratory microbiota: addressing clinical questions, informing chronic obstructive pulmonary disease. Thorax 2012;67:
clinical practice. Thorax 2015;70:74–81. 694–700.

986 American Journal of Respiratory and Critical Care Medicine Volume 193 Number 9 | May 1 2016
ORIGINAL ARTICLE

33. Lochner M, Ohnmacht C, Presley L, Bruhns P, Si-Tahar M, Sawa S, 52. Di Stefano A, Caramori G, Gnemmi I, Contoli M, Vicari C, Capelli A,
Eberl G. Microbiota-induced tertiary lymphoid tissues aggravate Magno F, D’Anna SE, Zanini A, Brun P, et al. T helper type 17-related
inflammatory disease in the absence of RORgamma t and LTi cells. cytokine expression is increased in the bronchial mucosa of stable
J Exp Med 2011;208:125–134. chronic obstructive pulmonary disease patients. Clin Exp Immunol
34. Wu HJ, Ivanov II, Darce J, Hattori K, Shima T, Umesaki Y, Littman DR, 2009;157:316–324.
Benoist C, Mathis D. Gut-residing segmented filamentous bacteria 53. Taraseviciene-Stewart L, Douglas IS, Nana-Sinkam PS, Lee JD, Tuder
drive autoimmune arthritis via T helper 17 cells. Immunity 2010;32: RM, Nicolls MR, Voelkel NF. Is alveolar destruction and emphysema
815–827. in chronic obstructive pulmonary disease an immune disease? Proc
35. Berer K, Mues M, Koutrolos M, Rasbi ZA, Boziki M, Johner C, Am Thorac Soc 2006;3:687–690.
Wekerle H, Krishnamoorthy G. Commensal microbiota and myelin 54. Sullivan AK, Simonian PL, Falta MT, Mitchell JD, Cosgrove GP, Brown
autoantigen cooperate to trigger autoimmune demyelination. Nature KK, Kotzin BL, Voelkel NF, Fontenot AP. Oligoclonal CD41 T cells in
2011;479:538–541. the lungs of patients with severe emphysema. Am J Respir Crit Care
36. Lee YK, Menezes JS, Umesaki Y, Mazmanian SK. Proinflammatory Med 2005;172:590–596.
T-cell responses to gut microbiota promote experimental autoimmune 55. Marsland BJ, Yadava K, Nicod LP. The airway microbiome and
encephalomyelitis. Proc Natl Acad Sci USA 2011;108:4615–4622. disease. Chest 2013;144:632–637.
37. Sajjan U, Ganesan S, Comstock AT, Shim J, Wang Q, Nagarkar DR, 56. Naik S, Bouladoux N, Wilhelm C, Molloy MJ, Salcedo R,
Zhao Y, Goldsmith AM, Sonstein J, Linn MJ, et al. Elastase- and Kastenmuller W, Deming C, Quinones M, Koo L, Conlan S, et al.
LPS-exposed mice display altered responses to rhinovirus infection. Compartmentalized control of skin immunity by resident
Am J Physiol Lung Cell Mol Physiol 2009;297:L931–L944. commensals. Science 2012;337:1115–1119.
38. Caporaso JG, Kuczynski J, Stombaugh J, Bittinger K, Bushman FD, 57. Molyneaux PL, Mallia P, Cox MJ, Footitt J, Willis-Owen SA, Homola D,
Costello EK, Fierer N, Peña AG, Goodrich JK, Gordon JI, et al. QIIME Trujillo-Torralbo MB, Elkin S, Kon OM, Cookson WO, et al.
allows analysis of high-throughput community sequencing data. Nat Outgrowth of the bacterial airway microbiome after rhinovirus
Methods 2010;7:335–336. exacerbation of chronic obstructive pulmonary disease. Am J Respir
39. Edgar RC. Search and clustering orders of magnitude faster than Crit Care Med 2013;188:1224–1231.
BLAST. Bioinformatics 2010;26:2460–2461. 58. Venkataraman A, Bassis CM, Beck JM, Young VB, Curtis JL,
40. DeSantis TZ, Hugenholtz P, Larsen N, Rojas M, Brodie EL, Keller K, Huffnagle GB, Schmidt TM. Application of a neutral community
Huber T, Dalevi D, Hu P, Andersen GL. Greengenes, a chimera- model to assess structuring of the human lung microbiome. MBio
checked 16S rRNA gene database and workbench compatible with 2015;6:6.
ARB. Appl Environ Microbiol 2006;72:5069–5072. 59. Bassis CM, Erb-Downward JR, Dickson RP, Freeman CM, Schmidt
41. R Development Core Team. R: a language and environment for TM, Young VB, Beck JM, Curtis JL, Huffnagle GB. Analysis of the
statistical computing. Vienna, Austria: R Foundation for Statistical upper respiratory tract microbiotas as the source of the lung and
Computing; 2008. gastric microbiotas in healthy individuals. MBio 2015;6:e00037.
42. Marsland BJ, Kurrer M, Reissmann R, Harris NL, Kopf M.
60. Shaw MH, Kamada N, Kim YG, Núñez G. Microbiota-induced IL-1b,
Nippostrongylus brasiliensis infection leads to the development of
but not IL-6, is critical for the development of steady-state TH17 cells
emphysema associated with the induction of alternatively activated
in the intestine. J Exp Med 2012;209:251–258.
macrophages. Eur J Immunol 2008;38:479–488.
61. Martin B, Hirota K, Cua DJ, Stockinger B, Veldhoen M. Interleukin-17-
43. Park H, Shin JW, Park SG, Kim W. Microbial communities in the upper
producing gammadelta T cells selectively expand in response to
respiratory tract of patients with asthma and chronic obstructive
pathogen products and environmental signals. Immunity 2009;31:
pulmonary disease. Plos One 2014;9:e109710.
321–330.
44. Engler K, Mühlemann K, Garzoni C, Pfahler H, Geiser T, von Garnier C.
62. Duan J, Chung H, Troy E, Kasper DL. Microbial colonization drives
Colonisation with Pseudomonas aeruginosa and antibiotic resistance
expansion of IL-1 receptor 1-expressing and IL-17-producing
patterns in COPD patients. Swiss Med Wkly 2012;142:w13509.
gamma/delta T cells. Cell Host Microbe 2010;7:140–150.
45. Lambiase A, Del Pezzo M, Raia V, Sepe A, Ferri P, Rossano F.
Chryseobacterium respiratory tract infections in patients with cystic 63. Trompette A, Gollwitzer ES, Yadava K, Sichelstiel AK, Sprenger N,
fibrosis. J Infect 2007;55:518–523. Ngom-Bru C, Blanchard C, Junt T, Nicod LP, Harris NL, et al. Gut
46. Zhang J, Chu S, Zhong X, Lao Q, He Z, Liang Y. Increased expression microbiota metabolism of dietary fiber influences allergic airway
of CD41IL-171 cells in the lung tissue of patients with stable disease and hematopoiesis. Nat Med 2014;20:159–166.
chronic obstructive pulmonary disease (COPD) and smokers. Int 64. Yadava K, Marsland BJ. Lymphoid follicles in chronic lung diseases.
Immunopharmacol 2013;15:58–66. Thorax 2013;68:597–598.
47. Podolin PL, Foley JP, Carpenter DC, Bolognese BJ, Logan GA, Long E 65. Kirkham PA, Caramori G, Casolari P, Papi AA, Edwards M, Shamji B,
III, Harrison OJ, Walsh PT. T cell depletion protects against alveolar Triantaphyllopoulos K, Hussain F, Pinart M, Khan Y, et al. Oxidative
destruction due to chronic cigarette smoke exposure in mice. Am J stress-induced antibodies to carbonyl-modified protein correlate
Physiol Lung Cell Mol Physiol 2013;304:L312–L323. with severity of chronic obstructive pulmonary disease. Am J Respir
48. Kurimoto E, Miyahara N, Kanehiro A, Waseda K, Taniguchi A, Ikeda G, Crit Care Med 2011;184:796–802.
Koga H, Nishimori H, Tanimoto Y, Kataoka M, et al. IL-17A is 66. Seys LJ, Verhamme FM, Schinwald A, Hammad H, Cunoosamy DM,
essential to the development of elastase-induced pulmonary Bantsimba-Malanda C, Sabirsh A, McCall E, Flavell L, Herbst R, et al.
inflammation and emphysema in mice. Respir Res 2013;14:5. Role of b cell-activating factor in chronic obstructive pulmonary
49. Eppert BL, Wortham BW, Flury JL, Borchers MT. Functional disease. Am J Respir Crit Care Med 2015;192:706–718.
characterization of T cell populations in a mouse model of chronic 67. Curtis JL, Freeman CM, Huffnagle GB. “B” for bad, beneficial, or both?
obstructive pulmonary disease. J Immunol 2013;190:1331–1340. Lung lymphoid neogenesis in chronic obstructive pulmonary
50. Anthony D, Seow HJ, Uddin M, Thompson M, Dousha L, Vlahos R, disease. Am J Respir Crit Care Med 2015;192:648–651.
Irving LB, Levy BD, Anderson GP, Bozinovski S. Serum amyloid A 68. Brandsma CA, Kerstjens HA, van Geffen WH, Geerlings M, Postma DS,
promotes lung neutrophilia by increasing IL-17A levels in the mucosa Hylkema MN, Timens W. Differential switching to IgG and IgA in
and gd T cells. Am J Respir Crit Care Med 2013;188:179–186. active smoking COPD patients and healthy controls. Eur Respir J
51. Chen K, Pociask DA, McAleer JP, Chan YR, Alcorn JF, Kreindler JL, 2012;40:313–321.
Keyser MR, Shapiro SD, Houghton AM, Kolls JK, et al. IL-17RA is 69. Murphy TF, Brauer AL, Grant BJ, Sethi S. Moraxella catarrhalis in
required for CCL2 expression, macrophage recruitment, and chronic obstructive pulmonary disease: burden of disease
emphysema in response to cigarette smoke. Plos One 2011;6: and immune response. Am J Respir Crit Care Med 2005;172:
e20333. 195–199.

Yadava, Pattaroni, Sichelstiel, et al.: Microbiota Sustains Chronic Pulmonary Inflammation 987

You might also like